Next Article in Journal
Secondary Metabolites from the Endoparasitic Nematophagous Fungus Harposporium anguillulae YMF 1.01751
Next Article in Special Issue
Viral Infection and Antiviral Treatments in Ocular Pathologies
Previous Article in Journal
The Scenario of Ticks and Tick-Borne Pathogens of Sheep on a Mediterranean Island
Previous Article in Special Issue
In Vitro and In Silico Analysis of the Inhibitory Activity of EGCG-Stearate against Herpes Simplex Virus-2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Novel Approach of Antiviral Drugs Targeting Viral Genomes

1
Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Korea
2
Novelgen Co., Ltd., R&D Center, 77, Changnyong-daero 256 Beon-gil, Yeongtong-gu, Suwon 16229, Gyeonggi-do, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2022, 10(8), 1552; https://doi.org/10.3390/microorganisms10081552
Submission received: 3 June 2022 / Revised: 25 July 2022 / Accepted: 29 July 2022 / Published: 31 July 2022
(This article belongs to the Special Issue Emerging Viruses and Antiviral Drugs)

Abstract

:
Outbreaks of viral diseases, which cause morbidity and mortality in animals and humans, are increasing annually worldwide. Vaccines, antiviral drugs, and antibody therapeutics are the most effective tools for combating viral infection. The ongoing coronavirus disease 2019 pandemic, in particular, raises an urgent need for the development of rapid and broad-spectrum therapeutics. Current antiviral drugs and antiviral antibodies, which are mostly specific at protein levels, have encountered difficulties because the rapid evolution of mutant viral strains resulted in drug resistance. Therefore, degrading viral genomes is considered a novel approach for developing antiviral drugs. The current article highlights all potent candidates that exhibit antiviral activity by digesting viral genomes such as RNases, RNA interference, interferon-stimulated genes 20, and CRISPR/Cas systems. Besides that, we introduce a potential single-chain variable fragment (scFv) that presents antiviral activity against various DNA and RNA viruses due to its unique nucleic acid hydrolyzing characteristic, promoting it as a promising candidate for broad-spectrum antiviral therapeutics.

1. Introduction

Viral infections pose a major risk to global health. The most recent case is the coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has infected more than 513 million people worldwide, with 6.2 million deaths as of May 2022 [1]. Other coronavirus strains cause life-threatening diseases such as SARS and the Middle East respiratory syndrome (MERS) [2,3]. Re-emerging virus infection, such as the Ebola virus, was reported in West Africa in 2014–2016, causing an outbreak with a case fatality rate of up to 50%, and the most recent case, which is still ongoing, has been reported in the Democratic Republic of Congo with a case fatality rate of 66% [4]. In April 2022, new Ebola virus cases were reported in the Mbandaka, Equateur Province of the Democratic Republic of Congo, indicating that the Ebola virus remains a major threat to global health [5]. On the other hand, viral infections caused by human immunodeficiency virus (HIV), hepatitis B virus (HBV), and hepatitis C virus (HCV) continue to pose a threat to global health. Globally, it is reported that approximately 1.7 million people have been newly infected with HIV, with 700,000 HIV-related deaths [6]. In addition, approximately 250 million and 71 million people have been reported to have chronic HBV and HCV, respectively, and the death toll caused by this viral hepatitis is estimated at approximately 1.34 million [7]. Flu caused by the influenza virus is also a global health issue yearly. In 2009, a global pandemic caused by the influenza A (H1N1) pdm09 virus resulted in more than 150,000–570,000 deaths worldwide, and it is currently circulating as a virus causing seasonal flu [8]. Avian influenza A (H5N1), another variant of influenza viruses, has become a concern since its first human infection in 1997 [9]. The viral epidemic and pandemic have caused many casualties throughout history, which are summarized in Table 1.
The viral life cycle consists of several major stages, including virus attachment to the membrane of the host cell, viral entry (i.e., endocytosis), viral uncoating, viral genome transcription and replication, viral proteins translation, viral assembly, and viral budding/release. The mechanisms of viral transcription and replication vary depending on their genome type (RNA/DNA; single- or double-stranded; and positive (+) or negative (−) polarity) [10]. Notably, regardless of the virus type, all viruses must release the viral genomes to the cytoplasm, produce mRNA, and replicate their genomes. Most DNA viruses transcribe their genome in the nucleus of the host cells and use the host enzymes for their DNA replication and gene expression; except the Poxvirus such as the Vaccinia virus, which can independently replicate in the cytoplasm because of its viral genome that encodes viral proteins responsible for transcription and replication processes [11]. The RNA viruses replicate their genome in the cytoplasm of the host cell with the help of RNA-dependent-RNA polymerase (RdRp). However, in retroviruses such as HIV, the RNA genome is reversely transcribed by RNA-dependent-DNA polymerase (RdDp) and subsequently delivered to the nucleus to integrate with the host genome [12,13]. Another example of RNA viruses replicating within the nucleus is influenza viruses [14].
The stages of the viral life cycle have become a common target for the development of antiviral drugs against viral or host cell proteins to inhibit viral replication [10,15]. Although numerous approaches against viral infection have been developed and approved, the fact that drug-resistant virus strains are increasing has made it difficult to use current drugs for treating viral diseases [16,17,18,19,20]. Therefore, in an effort to discover new drugs capable of combating a broad spectrum of viruses, we introduce a novel platform that directly targets viral genome degradation to inhibit viral replication. Here, to the best of our knowledge, we reviewed all candidates that show antiviral effects in the in vitro and in vivo tests of viral genome cleavage, including ribonucleases (RNases), interferon (IFN)-stimulated genes 20 (ISG20), RNA interference (RNAi), CRISPR/Cas systems, and a catalytic single-chain variable fragment 3D8 (3D8 scFv). They presented as the most straightforward and universal strategies for cleaving the viral genomes.

2. Antiviral Approach Targeting Viral Genomes

2.1. RNases

RNases, as a classical component of host defense, have been proposed as therapeutic candidates by catalyzing viral RNA. RNases, in addition to degrading viral RNA, play roles in IFN activation, viral replication inhibition, and apoptosis induction [21]. RNases have the potential to degrade viral genomes, but a major challenge is to limit host cell destruction. Numerous RNases exhibit antiviral properties against single-stranded RNA (ssRNA) viruses [22,23]. Here, we briefly described the RNase antiviral candidates promoted by viral RNA degradation. RNase A superfamily, EDN/RNase 2, cleaved the viral RNA against HIV-1 [24]. RNase L is induced by IFN to cleave cytoplasmic RNA through the OAS/RNase L signal pathway. RNase L is activated by 2′-5′ oligoadenylates (2–5A). OAS is activated by double-stranded RNA (dsRNA) binding to synthesize 2–5A. RNase L catalyzed viral RNAs of IAV [25] and WNV [26,27] in an in vitro experiment. Viral RNAs of HCV and DENV were digested by Regnase/MCPIP1 [28,29]. Recently, the antiviral RNases, such as OSA/RNase L have been well-characterized and exhibit multiple antiviral mechanisms. However, they present several challenges. There is incomplete cell protection against viruses via RNases, for instance, viruses escaping from RNases L have indicated the following in some cases: a neurotropic Theiler’s picornavirus produces a helper protein that inhibits RNases L [30], or DENV and IAV mRNAs escape RNase L-mediated decay, resulting in the production of viral protein [31]. RNases must be retained at the appropriate concentration to exhibit antiviral activity without exaggerated RNases cytotoxicity [32].

2.2. RNAi

RNAi is a cellular gene-silencing pathway that degrades the sequence-specific mRNA [33]. Since its discovery in 1998, RNAi has rapidly devolved as a therapeutic drug application with small interfering RNAs (siRNAs) being the most commonly used drugs. siRNAs have been widely studied against different viruses that degrade all types of viral genomes ds or ss DNA/RNA such as HIV [34,35], influenza virus [36,37], SARS-CoV [38,39], HBV [40,41], HPV [42], and WNV [43]. The FDA approved the first siRNA-based drug in 2018, followed by three more commercial drugs until now [44]. This was a significant step in siRNA research toward practicality. This indicated the potential of siRNAs as antiviral drugs in the future when some siRNA-based antiviral drugs entered the clinical phases, for instance, NucB1000 in phase I targets four HBV genes [45], ALN-RSV01 in phase II targets the nucleocapsid gene of RSV [46], pHIV7-shI-TAR-CCR5RZ in phase I targets multiple genes of HIV [33], TKM-Ebola in phase I targets multiple transcripts of Ebola virus [47]. The mechanism of siRNAs is as follows: after being taken up by the cell, numerous processes are performed by entering endosomes and following the endosomal escape. Then, siRNAs are released into the cytoplasm and taken up by the multiprotein RNA-induced silencing complex (RISC), and siRNAs guide the RISC complex to identify and cleave the targetable regions [44]. One of the barriers to its application is the delivery of siRNA to the target cells and cell penetration. In addition, siRNA technology has faced and overcome several challenges, such as poor stability, immunostimulation, off-target effects, viral escape due to mutations, and durable viral silencing to achieve sustainable antiviral therapy [33,44].

2.3. ISG20

IFN or dsRNA induces ISG20 expression and its presence in the cytoplasm and nucleus [48,49,50]. ISG20 is a 3′-5′ exonuclease that belongs to the DEDDh subgroup of the DEDD exonuclease superfamily [51]. ISG20 cleaved both ssRNA and DNA but showed a preference for RNA substrates over DNA by Met14 and Arg53 to accommodate hydrogen bonds with the 2′-OH group of UMP ribose [52,53]. Therefore, IGS20 exhibited greater antiviral activity to multiple RNA viruses than to DNA viruses [54] but failed to prevent SARS-CoV and DNA viruses, such as adenoviruses. Viral RNA degradation and direct or indirect inhibition of viral translation are the two proposed antiviral mechanisms of ISG20 (recently reviewed in [55]). The lack of viral inhibition by catalytically inactive ISG20 form-D94G supports the hypothesis that ISG20 exonuclease activity determines viral genome degradation. However, the specific action of viral genome degradation without damaging the host cells is yet to be investigated [52,56]. ISG20 has no apparent sequence specificity for viral genome degradation. However, in HBV infection, its overexpression does not cause cellular RNA degradation by the cellular cofactor YTHDF2. YTHDF2 is a cytoplasmic YT521-B homology domain-containing protein, the main group of m6A readers in the m6A pathway. The recruitment of ISG20 for RNA degradation through the binding of YTHDF2 to m6A modified the HBV pregenomic RNA. This study demonstrated that HBV RNA was selectively degraded by IGS20 [57,58,59,60].
Three factors regulate the selective binding of ISG20 to viral RNA of HBV: the appropriate specific binding site (the sequence GGACA of HBV ε loop), m6A modification, and the host factor, YTHDF2. Although this mechanism is specific to HBV, a similar scheme may be applied to other viral RNAs [57]. Most recent studies have shown the antiviral effect of ISG20 via the ectopic expression or generation of transgenic cell lines; therefore, it is necessary to focus on the mechanism underlying the intracellular delivery of ISG20 as an antiviral agent (Table 2).
The finding that ISG20 antiviral activity was not mediated by RNA degradation but rather by translation inhibition demonstrated the exonuclease-independent antiviral activities of ISG20. The overexpression of ISG20 correlated with the expression of IFIT1 preventing viral RNA translation with non-2′O-methylated 5′ caps [61]. The viral translation inhibition is mediated by ISG20 discrimination of self-nucleic acid from nonself [60]. Furthermore, ISG20 exhibited HBV replication inhibition via exonuclease-independent activities [56]. Obviously, ISG20 acquired multiple antiviral mechanisms, which prompted the development of a potential antiviral drug that is not limited to viral genome degradation.
Table 2. ISG20 exhibited an antiviral effect against viruses via viral genome degradation.
Table 2. ISG20 exhibited an antiviral effect against viruses via viral genome degradation.
ISG20 DerivationVirusGenomeReplicationGenusFamilyRef
Ectopic expressionHBVDouble-stranded, relaxed circular DNA (rcDNA)NucleusOrthohepadnavirusHepadnaviridae[56,57,58]
Ectopic expressionYFV(+)ssRNACytoplasmFlavivirusFlaviviridae[62]
Transgenic cell lineBVDV(+)ssRNACytoplasmPestivirusFlaviviridae
Transgenic cell lineHAV(+)ssRNACytoplasmHepatovirusPicornaviriade
Transgenic cell lineVSV(−)ssRNACytoplasmVesiculovirusRhabdoviridae[63]
Transgenic cell lineInfluenza(−)ssRNANucleusAlphainfluenzavirusOrthomyxoviridae
Transgenic cell lineEMCV(+)ssRNACytoplasmCardiovirusPicornaviriade
Transgenic cell lineWNV(+)ssRNACytoplasmFlavivirusFlaviviridae[64]
Transgenic cell lineDENV(+)ssRNACytoplasmFlavivirusFlaviviridae
Transgenic cell lineHCV(+)ssRNACytoplasmHepacivirusFlaviviridae[65]

2.4. CRISPR/Cas System

The clustered regularly interspaced short palindromic repeats (CRISPR)-Cas (CRISPR-associated proteins), known as the immune system of bacteria and archaea, has been a powerful tool for editing genes. Nevertheless, it has been developed as a CRISPR/Cas-based antiviral therapy, making it one of the potential candidates for viral treatment. In this section, we only focused on the feasibility of directly targeting the virus of CRISPR/Cas without modifying the host genome. CRISPR/Cas can be categorized into two main classes, with six types and several subtypes based on the effector Cas domain content [66,67]. Of these, class II Cas endonucleases have recently been a popular topic for antiviral studies, with the ability of dsDNA digestion of effectors, such as Cas9, Cas12, and ssRNA cutting of Cas13 [68]. The CRISPR/Cas system works on the principle that a single-guide RNA (sgRNA), after transcribing into small antisense CRISPR RNA (crRNA), guides the crRNA-Cas ribonucleoprotein complex, which eventually recognizes the specific sequences and destroys the viral targets (including intracellular viral genome and viral mRNAs) [67,68].
The CRISPR/Cas9 method is a promising tool for chronic diseases, as demonstrated by the studies shown in Table 3, which show decreased viral genome [69,70], declined latency reactivation [71,72,73], reduced GFP expression [71,72,73,74], and viral protein [72,74]. Within the six domains (REC I, REC II, Bridge Helix, PAM-interacting, HNH, and RuvC) of Cas9, the PAM-interacting domain binds to the sequence that matches its PAM after scanning the target DNA, and then the guide RNA (gRNA) complex melts the bases right upstream of PAM and complement them with the target on the gRNA. If this association occurs appropriately, RuvC and HNH-nuclease domains will cleave the target DNA after the third nucleotide upstream of PAM [66,75]. Similar to Cas9, Cas12 attacks dsDNA but has fewer off-target effects than Cas9. However, both require PAM with G-rich PAM for Cas9 and T-rich PAM for Cas12. The mechanism of Cas12 has some differences compared with that of Cas9 because Cas9 digests a target that is right next to PAM with the blunt-ended dsDNA breaks, but Cas12 exerts its effects downstream at some distance from PAM with sticky ends [76]. Although Cas9 can inhibit both DNA and RNA viruses by introducing DNA intermediates into the host cell, most of the viruses that infect humans are RNA viruses with more than 180 of 200 RNA viruses infecting humans [77,78,79]. Therefore, RNA-targeting CRISPR/Cas13 has been studied to address this issue. The Cas13 family contains at least four subtypes: Cas13a, b, c, and d. Unlike Cas9 and Cas12, Cas13 has a dual conserved higher eukaryote and prokaryote nucleotide-binding domain (HEPN) instead of the DNase domain (RuvC and HNH), as well as recognizes target RNA based on the PFS domain instead of the PAM domain [80]. Subsequently, target RNA is identified via PFS, and bound by gRNA, and Cas13 then cleaves the target RNA [80,81]. The first approach of Cas13 against SARS-CoV-2 in human cells (PACMAN-prophylactic antiviral CRISPR in human cells) used pan-coronavirus crRNAs; Abbott et al. discovered that six crRNAs could target 91% of the sequenced coronaviruses, leading to several other studies using Cas13-targeted SARS-CoV-2 and influenza virus, resulting in positive outcomes in vitro and in vivo [68,82,83]. Additionally, targeting positive-sense viruses results in the degradation of viral gene expression by Cas13 and the preparation of RNA for assembling new particles as well as viral RNA genome serving as templates for replication [68,82].
Apart from the apparent advantages, the main challenges include accurate delivery, high specificity, lower immunogenicity, and long-lasting effect on inoculated in vivo models. To date, there is consensus over whether Cas13 exhibits significant off-target effects. In most HIV research, no off-target effect has been reported, and the collateral damage is undetectable in many applications using Cas13s as the RNA targeting system in mammalian cells, animal models, and plants [75,84,85,86,87,88,89,90]. Nonetheless, the extent of the off-target effects indicated the differences based on the cell type and target RNA observed. For instance, Yuxi Ai et al. showed that the extension of the off-target effects of CRISPR/Cas13s limited their utility in eukaryotic cells [91]. Therefore, this notion remains a challenge limiting their clinical trial. Notably, Cas13 research design and interpretation should be carefully performed to achieve potential results [91].
To use the CRISPR/Cas system, sgRNAs must be designed and constructed based on highly conserved regions of the viral genome to avoid escape mutants and reduce the possibility of off-target effects. This is followed by cloning these sgRNAs into an expression plasmid or viral vector, and then expressed using lipofectamine transfection or lentiviral/adenoviral transduction [68].
Table 3. Viral genome-specific cleavage by CRISPR with different Cas effector.
Table 3. Viral genome-specific cleavage by CRISPR with different Cas effector.
CasExperimentTarget GeneVirusGenomeReplicationGenusFamilyRef.
Cas9WSL-gRp30 cellp30 gene (CP204L)ASFV (BA71V)dsDNACytoplasmAsfivirusAsfarviridae[92]
Vero, ICP0-complementing L7 cell line 27, TC620ICP0, ICP4, and ICP27 genesHSV-1dsDNANucleusSimplexvirusHerpesviridae[93]
Vero, 239T, and BALB/c miceUL7 genes[94]
Vero cellUL15, UL27, UL29, UL30, UL36, UL37, UL42, UL5, UL52, UL8, UL54, UL9, US3, and US8[95]
Vero cellUL8, UL29, and UL52[96]
297T, HaCaT, HaCaT IFNAR2-knockout, THP-1, primary mouse corneal stromal cell, and C57BL/6J miceUL8 and UL29 genes[97]
HEK293T, HeLa, and Jurkat c5 and c19 cellsTLRHIV-1(−)ssRNAReverse transcription in cytoplasm
Replication in nucleus
LentivirusRetrovirus[82]
CHME5 cell, HeLa-derived TZM-bI cells, promonocytic U-937 cell subclone U1TLR-U3[83]
Transgenic mice5′-LTR and Gag gene[98]
Human T-lymphoid cell, Jurkat 2D10, PBMCsLTR-U3[99]
NRG miceLTR[71]
HEK293T, Jurkat C11, and TZM-bl cellsLTR7, LTR8, and structural region (env5, vif2, rev3, gag8, pol6, and pol7)[100]
HEK293TLTR, gag, and pol[84]
Tg26 transgenic mice, BLT mice, NCr nude miceLTR, gag, and pol[101]
HEK293FT, primary human monocytesLTR, gag, env, ref, tat[102]
Cas12aHEK293T cellLTR, gag, env, pol, tat, rev, nef, vpr[70]
Cas13aHEK293T cell, HEK293 cellLTR, gag, tat, and rev[103]
Cas9HepG2.2.15 cellHBV DNA sequencesHBVDouble-stranded, relaxed circular DNA (rcDNA)NucleusOrthohepadnavirusHepadnaviridae[72]
HepG2 cell, and Balb/c miceConserved regions of HBV[74]
Huh7 cell, HepG2.2.15 cell, and Balb/c mice20 nucleotide HBV DNA sequences[104]
HepG2 cellConserved regions of HBV[105]
Huh7 cell, and C57BL/6 miceConserved regions of HBV[106]
PK-15 cellUL30PRVdsDNANucleusVaricellovirusHerpesviridae[73]
Vero cellEssential and nonessential genes[107]
Vero cellEBNA1, OriPEBVDNANucleusLymphocryptovirusHerpesviridae[95]
Vero cellUL54, UL44, UL57, UL70, UL105, UL86, and UL84HCMVDNANucleusCytomegalovirusHerpesviridae[95]
Cas9Hela cell, Caski, HEK293T, Jurkat, Hela-FLAG16E7MYC cell, and Rag1 miceE6 and E7HPVdsDNANucleusAlphapapillomavirusPapovaviridae[108]
SiHa, C33-A, and BALB/c nude miceE6 and E7[109]
SiHa cell, and nude miceE7[110]
Hela, HCS-2, SKG-I, 293, and BALB/c nude miceE6[111]
Hela, 293T, and SiHa cellE6 and E7HPVdsDNANucleusAlphapapillomavirusPapovaviridae[112]
Cas12aBmN-SWU1 cell, and transgenic silkwormei-1 geneBmNPVdsDNANucleusAlphabaculovirusBaculovirudae[69]
Cas13aMicePB1 and PB2 genes of influenzaIAV-H1N1 (A/WSN/33)(−)ssRNANucleusAlphainfluenzavirusOrthomyxoviridae[89]
MDCK cellConserved regions of H1N1IAV-H1N1 (A/Puerto Rico/8/1934)(−)ssRNANucleusAlphainfluenzavirusOrthomyxoviridae[81]
A549 cellConserved regions of H1N1IAV-H1N1(−)ssRNANucleusAlphainfluenzavirusOrthomyxoviridae[77]
HamstersReplicase and nucleocapsid genes of SARSSARS-CoV2(+)ssRNACytoplasmBetacoronavirusCoronaviridae[89]
HepG2 cell, and AT2 cellS geneSARS-CoV2[113]
A549 cell, and HEK293T cellRdRp (ORF1ab) and N geneSARS-CoV2(+)ssRNACytoplasmBetacoronavirusCoronaviridae[77]
-Replicase and transcriptase (ORFab) and S geneSARS-CoV2(+)ssRNACytoplasmBetacoronavirusCoronaviridae[114]
HEK293FT cellConserved regions of LCMVWild type-LCMV Armstrong(−)ssRNACytoplasmMammarenavirusArenaviridae[81]
HEK293FT cellConserved regions of VSVVSV(−)ssRNACytoplasmVesiculovirusRhabdoviridae[81]
HEK293T, HEK293FT, and MARC-145 cellORF5 and ORF7PRRSV(+)ssRNACytoplasmPorartevirusArteriviridae[115]

2.5. 3D8 Single-Chain Variable Fragment (3D8 scFv)

A monoclonal antibody (mAb) named 3D8 was discovered and isolated as an anti-DNA Abs from the spleen cells of MRL-lpr/lpr mice, an autoimmune-prone mouse model that resembles human systemic lupus erythematosus. Sequentially, a recombinant 3D8 single-chain variable fragment (3D8 scFv), 27kD, was generated in the formation of the heavy chain variable single domain (VH) connecting the light chain variable single domain (VL) via a flexible (Glycine4-Serine1)3 linker [116]. The 3D8 scFv protein can bind and hydrolyze nonspecific on both DNA (dsDNA, ssDNA) and RNA (dsRNA, ssRNA) in the presence of Mg2+ [117,118]. Although methylated and histone-bound DNA were not cleaved by 3D8 scFv, viral RNA hydrolyzed in vRNP form was recently reported [119,120]. The 3D8 scFv protein can be obtained from different systems, and the protein retains functional activities such as E. coli bacterial expression system [116,117], Lactobacillus paracasei [121], or in vegetatively reproductive kalanchoe pinnata via planta transformation [122]. However, human embryonic kidney 293f (HEK293f)-derived 3D8-His scFvs lost all DNA-hydrolyzing activity but retained its DNA-binding activity [123]. Unlike other cell-penetrating anti-DNA, Abs reported so far eventually traffic toward the nucleus. The 3D8 scFv can penetrate cells via caveolae/lipid raft endocytosis, which is mediated by heparan sulfate proteoglycans (HSPGs) and chondroitin sulfate proteoglycans (CSPGs) that function as endocytic receptors on the cell surface [124] and accumulate and remain up to 48 h in the cytosol without further translocation into endosomes, lysosomes, endoplasmic reticulum, Golgi, or nucleus [125]. Organ penetration of 3D8 scFv in vivo was revealed by its localization in the intestinal villi and lamina propria [121], epithelial cells, medium diameter bronchi and alveoli [126], and muscle, liver, lung, and brain tissues [118]. Furthermore, 3D8 scFv was maintained in the lung for 12 h by IN injection; in the liver, kidney, plasma, and lung for up to 6 h by IV or IP injection. An increase in the injection frequency extended 3D8 scFv retention time in the organ. In all transgenic chicken, mice, plants, or stable cell lines harboring the 3D8 scFv gene, the protein concentration was expressed as too low to be detectable, which is sufficient for conferring antiviral effects without incurring damage to host DNA/RNA. A sufficient 3D8 scFv dose is critical to target only viral DNA and RNA but not their host genetic material. Nucleic acid hydrolyzing 3D8 scFv treatment exerted some cytotoxic effects on cells because of its ability to hydrolyze cellular RNAs at high concentrations [125]. A treatment with 5 μM of 3D8 scFv caused no cytotoxicity for 48 h in cells [127]. There were no dead mice found for up to 5 d at 20, 50 μg [118,126], or up to 300 μg of 3D8 scFv injection [120]. Nucleic acid-hydrolyzing catalytic and cell penetration or organ distribution activities of 3D8 scFv has prompted its investigation in multiple antiviral applications. Interestingly, 3D8 scFv has shown antiviral activity against a broad spectrum of DNA and RNA viruses in both in vitro and in vivo experiments (Table 4). As a result, 3D8 scFv can be applied as a feed additive. Assessment for the use of purified 3D8 scFv as a feed additive according to the concept of substantial equivalence was confirmed, in which oral injection of E. coli 3D8 scFv was excreted from mice within the normal digestive transit time of the animals without colonization to the gastrointestinal tract [128]. Moreover, the administration of Lactobacillus salivarius expressing 3D8 scFv or Lactobacillus reuteri harboring 3D8 scFv as a feed additive enhances growth performance, immune homeostasis, and gut microbiota of chickens [129,130]. A metagenomic analysis revealed that probiotic Lactobacillus paracasei expressing 3D8 scFv enhances the probiotic activities in mice without any observable side effects [131]. 3D8scFv that originated from mouse (m3D8 scFv) were humanized (h3D8 scFv) or chickenized (ck3D8 scFv). Both h3D8 scFv and ck3D8 scFv retained the biochemical properties of m3D8 scFv as well as the structure. Importantly, ck3D8 scFv expressed lower immunogenicity than m3D8 scFv in chickens [132,133].

3. Discussion

Since the approval of idoxuridin, the first approved antiviral drug for the treatment of hepatitis B in 1963, many approved antiviral drugs have been commercially available for use. These antiviral drugs have a mode of action wherein they target the major stage of the viral life cycle to interfere with its replication process, as summarized in Figure 1A [15]. Based on the understanding of virology, antiviral drugs have been developed and are concentrated on two different approaches: targeting the host cell factors or the viral life cycle at protein levels [11].
In this review, we take the opportunity to propose a novel approach for antiviral drug degradation of the viral genome, which can be applied against various broad-spectrum viruses (Figure 1). From a perspective used as an antiviral approach, it is necessary to ensure that these candidates are correctly delivered to the target. Antiviral activity of these candidates in the in vitro and in vivo experiments was obtained via the ectopic expression by transient transfection or generation of transgenic cell lines, and animals. Undoubtedly, the use of suitable delivery vehicle tools by transfection, viral genome catalytic candidates, RNases, RNAi, and ISG20 or CRISPR/Cas, could expand the antiviral activity. Furthermore, 3D8 scFv was shown to penetrate cells, and it was localized in the cell cytoplasm via caveolae/lipid raft-mediated endocytosis [124,125] (Figure 1B). Each candidate can target different viral genomes stages, as indicated in (Figure 1C). As described, the presence of these candidates in the cytoplasm can digest the viral genome at the (1) releasing step, (2) viral mRNA transcription, or (3) replicated viral genomes for viral assembly.
The discovery of broad-spectrum antiviral agents, or “one drug, multiple viruses,” have been developed to protect us from various unknown and emerging viruses. However, a major challenge in developing antiviral drugs is a viral mutation, such as antigenic drift and antigenic shift in influenza virus, which causes the emergence of novel and drug-resistant strains of viruses. Other problems include the time and cost required to develop a new drug. Therefore, we aimed to introduce potential universal antiviral therapeutic targeting to digest viral genomes, a direct approach to inhibit viral replication. Notably, each candidate presents specific disadvantages that prompted further studies. For example, the unspecific cleavage site of RNases, ISG20, and 3D8 scFv encountered difficulty in discriminating viral genome and host cell nucleic acid, leading to the death of infected cells. Although many studies reported that a certain amount of 3D8 scFv demonstrated antiviral activity to various viruses and was not harmful to host cells, the selectively targeted viral genome requires further modification. Additionally, RNases and ISG20 exhibited antiviral activity not only by degrading the viral genome but also by relating to other pathways. RNases regulate the host immune, the formation of stress granules (SGs), inducing autophagy, and triggering apoptosis [21,22]. ISG20 inhibited HBV replication by exonuclease-dependent and -independent activities [56]. There are several challenges in RNAi application, such as, delivery to target cells, stability, and virus evolution [33,44]. In the case of the CRISPR/Cas system recognizing specific in the viral genome, CRISPR/Cas9, Cas13 decreases the cleavage activity in the existence of two or more mismatches between target RNA and crRNA [142]. There have been several challenges, such as delivery route, standard viral delivery vectors, sgRNA design, off-target effects, safety, and immunogenicity, which need to be overcome to achieve the further success of the CRISPR/Cas: approach to the supply chain, clinical development, and commercialization, with the researchers’ efforts, CRISPR/Cas has been refined over time. However, it could offer a powerful antiviral treatment against emerging, re-emerging viruses in the near future.
In conclusion, we introduced a novel approach exhibiting direct and universal antiviral therapy by cleavage of the viral genomes, regardless of the type of viruses. RNases, ISG20, a catalytic nucleic acid hydrolyzing 3D8 scFv, RNAi, and CRISPR/Cas systems that target viral genome degradation have been shown to exhibit antiviral activity. We reviewed all aspects of the candidates, and proposed insight mechanisms for each one to be considered as antiviral therapy.

Author Contributions

S.L. and P.T.H. determined the scope and conceptualized the review; S.L., P.T.H. and Q.X.T.L. outlined the draft; P.T.H., Q.X.T.L. and R.Q.A. drafted the manuscript; Y.L., T.T.B.V. and T.K. provided critical feedback on the outline of the review. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the Novelgen Company (Project NO. S-2018-1158-000).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We thank Songhee-Han for the illustrated figure.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Coronavirus Disease (COVID-19) Dashboard; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  2. WHO. Summary of Probable SARS Cases with Onset of Illness from 1 November 2002 to 31 July 2003; World Health Organization: Geneva, Switzerland, 2015. [Google Scholar]
  3. WHO. Middle East Respiratory Syndrome Coronavirus (MERS-CoV)—Saudi Arabia; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  4. WHO. Ebola in Democratic Republic of Congo—North Kivu, Ituri 2018–2020; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  5. WHO. Ebola Virus Disease-Democratic Republic of Congo; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  6. WHO. Latest HIV Estimates and Updates on HIV Policies Uptake, July 2020; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  7. WHO. Global Hepatitis Report, 2017; WHO: Geneva, Switzerland, 2017. [Google Scholar]
  8. Dawood, F.S.; Iuliano, A.D.; Reed, C.; Martin, I.; Meltzer, D.K. Estimated global mortality associated with the first 12 months of 2009 pandemic influenza A H1N1 virus circulation: A modelling study. Lancet Infect. Dis. 2012, 12, 687–695. [Google Scholar] [CrossRef] [Green Version]
  9. WHO. Avian Influenza A (H5N1)—United States of America; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  10. Tompa, D.R.; Immanuel, A.; Srikanth, S.; Kadhirvel, S. Trends and strategies to combat viral infections: A review on FDA approved antiviral drugs. Int. J. Biol. Macromol. 2021, 172, 524–541. [Google Scholar] [CrossRef]
  11. Reyes, E.D.; Kulej, K.; Pancholi, N.J.; Akhta, L.N.; Avgousti, D.C.; Kim, E.U.; Bricker, D.K.; Spruce, L.A.; Koniski, S.A.; Seeholzer, S.H.; et al. Identifying Host Factors Associated with DNA Replicated During Virus Infection. Mol. Cell. Proteom. 2017, 16, 2079–2097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Boersma, S.; Rabouw, H.H.; Bruurs, L.J.M.; Pavlovic, T.; Vliet, A.L.W.; Beumer, J.; Clevers, H.; Kuppeveld, F.J.M.; Tanenbaum, M.E. Translation and Replication Dynamics of Single RNA Viruses. Cell 2020, 183, 1930–1945. [Google Scholar] [CrossRef] [PubMed]
  13. Lu, L.; Su, S.; Yang, H.; Jiang, S. Antivirals with common targets against highly pathogenic viruses. Cell 2021, 184, 1604–1620. [Google Scholar] [CrossRef]
  14. Dou, D.; Revol, R.; Östbye, H.; Wang, H.; Daniels, R. Influenza A Virus Cell Entry, Replication, Virion Assembly and Movement. Front. Immunol. 2018, 9, 1581. [Google Scholar] [CrossRef] [PubMed]
  15. Clercq, E.D.; Li, G. Approved Antiviral Drugs over the Past 50 Years. Clin. Microbiol. Rev. 2016, 29, 695–747. [Google Scholar] [CrossRef] [Green Version]
  16. Strasfeld, L.; Chou, S. Antiviral drug resistance: Mechanisms and clinical implications. Infect. Dis. Clin. N. Am. 2010, 24, 413–437. [Google Scholar] [CrossRef] [Green Version]
  17. Lampejo, T. Influenza and antiviral resistance: An overview. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 1201–1208. [Google Scholar] [CrossRef] [PubMed]
  18. Van Poelvoorde, L.A.E.; Saelens, X.; Thomas, I.; Roosens, N.H. Next-Generation Sequencing: An Eye-Opener for the Surveillance of Antiviral Resistance in Influenza. Trends Biotechnol. 2020, 38, 360–367. [Google Scholar] [CrossRef] [Green Version]
  19. Kumar, M.; Kuroda, K.; Dhangar, K.; Mazumder, P.; Sonne, C.; Rinklebe, J.; Kitajima, M. Potential Emergence of Antiviral-Resistant Pandemic Viruses via Environmental Drug Exposure of Animal Reservoirs. Environ. Sci. Technol. 2020, 54, 8503–8505. [Google Scholar] [CrossRef] [PubMed]
  20. Durmuş, S.; Ülgen, K. Comparative interactomics for virus-human protein-protein interactions: DNA viruses versus RNA viruses. FEBS Open Bio 2017, 7, 96–107. [Google Scholar] [CrossRef] [PubMed]
  21. Silverman, R.H. Viral encounters with 2’,5’-oligoadenylate synthetase and RNase L during the interferon antiviral response. J. Virol. 2007, 81, 12720–12729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Li, J.; Boix, E. Host Defence RNases as Antiviral Agents against Enveloped Single Stranded RNA Viruses. Virulence 2021, 12, 444–469. [Google Scholar] [CrossRef] [PubMed]
  23. Ilinskaya, O.N.; Mahmud, R.S. Ribonucleases as antiviral agents. Mol. Biol. 2014, 48, 615–623. [Google Scholar] [CrossRef] [PubMed]
  24. Lee-Huang, S.; Huang, P.L.; Sun, Y.; Huang, P.L.; Kung, H.F.; Blithe, D.L.; Chen, H.C. Lysozyme and RNases as anti-HIV components in beta-core preparations of human chorionic gonadotropin. Proc. Natl. Acad. Sci. USA 1999, 96, 2678–2681. [Google Scholar] [CrossRef] [Green Version]
  25. Cooper, D.A.; Banerjee, S.; Chakrabarti, A.; García-Sastre, A.; Hesselberth, J.R.; Silverman, R.H.; Barton, D.J. RNase L targets distinct sites in influenza A virus RNAs. J. Virol. 2015, 89, 2764–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Scherbik, S.V.; Paranjape, J.M.; Stockman, B.M.; Silverman, R.H.; Brinton, M.A. RNase L plays a role in the antiviral response to West Nile virus. J. Virol. 2006, 80, 2987–2999. [Google Scholar] [CrossRef] [Green Version]
  27. Samuel, M.A.; Whitby, K.; Keller, B.C.; Marri, A.; Barchet, W.; Williams, B.R.; Silverman, R.H.; Gale, M., Jr.; Diamond, M.S. PKR and RNase L contribute to protection against lethal West Nile Virus infection by controlling early viral spread in the periphery and replication in neurons. J. Virol. 2006, 80, 7009–7019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Lin, R.J.; Chien, H.L.; Lin, S.Y.; Chang, B.L.; Yu, H.P.; Tang, W.C.; Lin, Y.L. MCPIP1 ribonuclease exhibits broad-spectrum antiviral effects through viral RNA binding and degradation. Nucleic Acids Res. 2013, 41, 3314–3326. [Google Scholar] [CrossRef] [Green Version]
  29. Lin, R.J.; Chu, J.S.; Chien, H.L.; Tseng, C.H.; Ko, P.C.; Mei, Y.Y.; Tang, W.C.; Kao, Y.T.; Cheng, H.Y.; Liang, Y.C.; et al. MCPIP1 suppresses hepatitis C virus replication and negatively regulates virus-induced proinflammatory cytokine responses. J. Immunol. 2014, 193, 4159–4168. [Google Scholar] [CrossRef] [Green Version]
  30. Sorgeloos, F.; Jha, B.K.; Silverman, R.H.; Michiels, T. Evasion of Antiviral Innate Immunity by Theiler’s Virus L* Protein through Direct Inhibition of RNase L. PLoS Pathog. 2013, 9, e1003474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Burke, J.M.; Gilchrist, A.R.; Sawyer, S.L.; Parker, R. RNase L limits host and viral protein synthesis via inhibition of mRNA export. Sci. Adv. 2021, 7, eabh2479. [Google Scholar] [CrossRef] [PubMed]
  32. Makarov, A.A.; Ilinskaya, O.N. Cytotoxic ribonucleases: Molecular weapons and their targets. FEBS Lett. 2003, 540, 15–20. [Google Scholar] [CrossRef]
  33. Qureshi, A.; Tantray, V.G.; Kirmani, A.R.; Ahangar, A.G. A review on current status of antiviral siRNA. Rev. Med. Virol. 2018, 28, e1976. [Google Scholar] [CrossRef]
  34. Singh, S.K. RNA interference and its therapeutic potential against HIV infection. Expert Opin. Biol. Ther. 2008, 8, 449–461. [Google Scholar] [CrossRef]
  35. Rossi, J. RNAi as a treatment for HIV-1 infection. BioTechniques 2006, 40, S25–S29. [Google Scholar] [CrossRef]
  36. Ge, Q.; McManus, M.T.; Nguyen, T.; Shen, C.-H.; Sharp, P.A.; Eisen, H.N.; Chen, J. RNA interference of influenza virus production by directly targeting mRNA for degradation and indirectly inhibiting all viral RNA transcription. Proc. Natl. Acad. Sci. USA 2003, 100, 2718–2723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Nakazawa, M.; Kadowaki, S.-e.; Watanabe, I.; Kadowaki, Y.; Takei, M.; Fukuda, H. PA subunit of RNA polymerase as a promising target for anti-influenza virus agents. Antivir. Res. 2008, 78, 194–201. [Google Scholar] [CrossRef]
  38. Shi, Y.; Yang, D.H.; Xiong, J.; Jia, J.; Huang, B.; Jin, Y.X. Inhibition of genes expression of SARS coronavirus by synthetic small interfering RNAs. Cell Res. 2005, 15, 193–200. [Google Scholar] [CrossRef] [Green Version]
  39. Meng, B.; Lui, Y.-w.; Meng, S.; Cao, C.; Hu, Y. Identification of effective siRNA blocking the expression of SARS viral envelope E and RDRP genes. Mol. Biotechnol. 2006, 33, 141–148. [Google Scholar] [CrossRef]
  40. Konishi, M.; Wu, C.H.; Wu, G.Y. Inhibition of HBV replication by siRNA in a stable HBV-producing cell line. Hepatology 2003, 38, 842–850. [Google Scholar] [CrossRef]
  41. Chen, Y.; Mahato, R.I. siRNA pool targeting different sites of human hepatitis B surface antigen efficiently inhibits HBV infection. J. Drug Target. 2008, 16, 140–148. [Google Scholar] [CrossRef] [PubMed]
  42. Bousarghin, L.; Touzé, A.; Gaud, G.; Iochmann, S.; Alvarez, E.; Reverdiau, P.; Gaitan, J.; Jourdan, M.-L.; Sizaret, P.-Y.; Coursaget, P. Inhibition of cervical cancer cell growth by human papillomavirus virus-like particles packaged with human papillomavirus oncoprotein short hairpin RNAs. Mol. Cancer Ther. 2009, 8, 357–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kumar, P.; Lee, S.K.; Shankar, P.; Manjunath, N. A single siRNA suppresses fatal encephalitis induced by two different flaviviruses. PLoS Med. 2006, 3, e96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mehta, A.; Michler, T.; Merkel, O.M. siRNA Therapeutics against Respiratory Viral Infections-What Have We Learned for Potential COVID-19 Therapies? Adv. Healthc Mater. 2021, 10, e2001650. [Google Scholar] [CrossRef]
  45. Gish, R.G.; Satishchandran, C.; Young, M.; Pachuk, C. RNA Interference and Its Potential Applications to Chronic HBV Treatment: Results of a Phase I Safety and Tolerability Study. Antivir. Ther. 2011, 16, 547–554. [Google Scholar] [CrossRef] [Green Version]
  46. DeVincenzo, J.; Lambkin-Williams, R.; Wilkinson, T.; Cehelsky, J.; Nochur, S.; Walsh, E.; Meyers, R.; Gollob, J.; Vaishnaw, A. A randomized, double-blind, placebo-controlled study of an RNAi-based therapy directed against respiratory syncytial virus. Proc. Natl. Acad. Sci. USA 2010, 107, 8800–8805. [Google Scholar] [CrossRef] [Green Version]
  47. Choi, J.H.; Croyle, M.A. Emerging targets and novel approaches to Ebola virus prophylaxis and treatment. BioDrugs 2013, 27, 565–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Espert, L.; Rey, C.; Gonzalez, L.; Degols, G.; Chelbi-Alix, M.K.; Mechti, N.; Gongora, C. The exonuclease ISG20 is directly induced by synthetic dsRNA via NF-kappaB and IRF1 activation. Oncogene 2004, 23, 4636–4640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Gongora, C.; David, G.; Pintard, L.; Tissot, C.; Hua, T.D.; Dejean, A.; Mechti, N. Molecular cloning of a new interferon-induced PML nuclear body-associated protein. J. Biol. Chem. 1997, 272, 19457–19463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Imaizumi, T.; Mechti, N.; Matsumiya, T.; Sakaki, H.; Kubota, K.; Yoshida, H.; Kimura, H.; Satoh, K. Expression of interferon-stimulated gene 20 in vascular endothelial cells. Microbiol. Immunol. 2008, 52, 30–35. [Google Scholar] [CrossRef]
  51. Moser, M.J.; Holley, W.R.; Chatterjee, A.; Mian, I.S. The proofreading domain of Escherichia coli DNA polymerase I and other DNA and/or RNA exonuclease domains. Nucleic Acids Res. 1997, 25, 5110–5118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Nguyen, L.H.; Espert, L.; Mechti, N.; Wilson, D.M., 3rd. The human interferon- and estrogen-regulated ISG20/HEM45 gene product degrades single-stranded RNA and DNA in vitro. Biochemistry 2001, 40, 7174–7179. [Google Scholar] [CrossRef] [PubMed]
  53. Horio, T.; Murai, M.; Inoue, T.; Hamasaki, T.; Tanaka, T.; Ohgi, T. Crystal structure of human ISG20, an interferon-induced antiviral ribonuclease. FEBS Lett. 2004, 577, 111–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zheng, Z.; Wang, L.; Pan, J. Interferon-stimulated gene 20-kDa protein (ISG20) in infection and disease: Review and outlook. Intractable Rare Dis. Res. 2017, 6, 35–40. [Google Scholar] [CrossRef] [Green Version]
  55. Yang, E.; Li, M.M.H. All About the RNA: Interferon-Stimulated Genes That Interfere With Viral RNA Processes. Front. Immunol. 2020, 11, 605024. [Google Scholar] [CrossRef]
  56. Leong, C.R.; Funami, K.; Oshiumi, H.; Mengao, D.; Takaki, H.; Matsumoto, M.; Aly, H.H.; Watashi, K.; Chayama, K.; Seya, T. Interferon-stimulated gene of 20 kDa protein (ISG20) degrades RNA of hepatitis B virus to impede the replication of HBV in vitro and in vivo. Oncotarget 2016, 7, 68179–68193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Imam, H.; Kim, G.W.; Mir, S.A.; Khan, M.; Siddiqui, A. Interferon-stimulated gene 20 (ISG20) selectively degrades N6-methyladenosine modified Hepatitis B Virus transcripts. PLoS Pathog. 2020, 16, e1008338. [Google Scholar] [CrossRef] [PubMed]
  58. Imam, H.; Khan, M.; Gokhale, N.S.; McIntyre, A.B.R.; Kim, G.W.; Jang, J.Y.; Kim, S.J.; Mason, C.E.; Horner, S.M.; Siddiqui, A. N6-methyladenosine modification of hepatitis B virus RNA differentially regulates the viral life cycle. Proc. Natl. Acad. Sci. USA 2018, 115, 8829–8834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Zaccara, S.; Jaffrey, S.R. A Unified Model for the Function of YTHDF Proteins in Regulating m(6)A-Modified mRNA. Cell 2020, 181, 1582–1595.e18. [Google Scholar] [CrossRef] [PubMed]
  60. Wu, N.; Nguyen, X.N.; Wang, L.; Appourchaux, R.; Zhang, C.; Panthu, B.; Gruffat, H.; Journo, C.; Alais, S.; Qin, J.; et al. The interferon stimulated gene 20 protein (ISG20) is an innate defense antiviral factor that discriminates self versus non-self translation. PLoS Pathog. 2019, 15, e1008093. [Google Scholar] [CrossRef] [PubMed]
  61. Weiss, C.M.; Trobaugh, D.W.; Sun, C.; Lucas, T.M.; Diamond, M.S.; Ryman, K.D.; Klimstra, W.B. The Interferon-Induced Exonuclease ISG20 Exerts Antiviral Activity through Upregulation of Type I Interferon Response Proteins. mSphere 2018, 3, e00209-18. [Google Scholar] [CrossRef] [Green Version]
  62. Zhou, Z.; Wang, N.; Woodson, S.E.; Dong, Q.; Wang, J.; Liang, Y.; Rijnbrand, R.; Wei, L.; Nichols, J.E.; Guo, J.T.; et al. Antiviral activities of ISG20 in positive-strand RNA virus infections. Virology 2011, 409, 175–188. [Google Scholar] [CrossRef] [Green Version]
  63. Espert, L.; Degols, G.; Gongora, C.; Blondel, D.; Williams, B.R.; Silverman, R.H.; Mechti, N. ISG20, a new interferon-induced RNase specific for single-stranded RNA, defines an alternative antiviral pathway against RNA genomic viruses. J. Biol. Chem. 2003, 278, 16151–16158. [Google Scholar] [CrossRef] [Green Version]
  64. Jiang, D.; Weidner, J.M.; Qing, M.; Pan, X.B.; Guo, H.; Xu, C.; Zhang, X.; Birk, A.; Chang, J.; Shi, P.Y.; et al. Identification of five interferon-induced cellular proteins that inhibit west nile virus and dengue virus infections. J. Virol. 2010, 84, 8332–8341. [Google Scholar] [CrossRef] [Green Version]
  65. Jiang, D.; Guo, H.; Xu, C.; Chang, J.; Gu, B.; Wang, L.; Block, T.M.; Guo, J.T. Identification of three interferon-inducible cellular enzymes that inhibit the replication of hepatitis C virus. J. Virol. 2008, 82, 1665–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. van der Oost, J.; Westra, E.R.; Jackson, R.N.; Wiedenheft, B. Unravelling the structural and mechanistic basis of CRISPR-Cas systems. Nat. Rev. Microbiol. 2014, 12, 479–492. [Google Scholar] [CrossRef]
  67. Hille, F.; Charpentier, E. CRISPR-Cas: Biology, mechanisms and relevance. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2016, 371, 20150496. [Google Scholar] [CrossRef] [PubMed]
  68. Baddeley, H.J.E.; Isalan, M. The Application of CRISPR/Cas Systems for Antiviral Therapy. Front. Genome Ed. 2021, 3, 745559. [Google Scholar] [CrossRef] [PubMed]
  69. Dong, Z.; Qin, Q.; Hu, Z.; Zhang, X.; Miao, J.; Huang, L.; Chen, P.; Lu, C.; Pan, M. CRISPR/Cas12a Mediated Genome Editing Enhances Bombyx mori Resistance to BmNPV. Front. Bioeng. Biotechnol. 2020, 8, 841. [Google Scholar] [CrossRef] [PubMed]
  70. Gao, Z.; Fan, M.; Das, A.T.; Herrera-Carrillo, E.; Berkhout, B. Extinction of all infectious HIV in cell culture by the CRISPR-Cas12a system with only a single crRNA. Nucleic Acids Res. 2020, 48, 5527–5539. [Google Scholar] [CrossRef]
  71. Bella, R.; Kaminski, R.; Mancuso, P.; Young, W.-B.; Chen, C.; Sariyer, R.; Fischer, T.; Amini, S.; Ferrante, P.; Jacobson, J.M.; et al. Removal of HIV DNA by CRISPR from Patient Blood Engrafts in Humanized Mice. Mol. Ther. -Nucleic Acids 2018, 12, 275–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ramanan, V.; Shlomai, A.; Cox, D.B.T.; Schwartz, R.E.; Michailidis, E.; Bhatta, A.; Scott, D.A.; Zhang, F.; Rice, C.M.; Bhatia, S.N. CRISPR/Cas9 cleavage of viral DNA efficiently suppresses hepatitis B virus. Sci. Rep. 2015, 5, 10833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Peng, Z.; Ouyang, T.; Pang, D.; Ma, T.; Chen, X.; Guo, N.; Chen, F.; Yuan, L.; Ouyang, H.; Ren, L. Pseudorabies virus can escape from CRISPR-Cas9-mediated inhibition. Virus Res. 2016, 223, 197–205. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, X.; Hao, R.; Chen, S.; Guo, D.; Chen, Y. Inhibition of hepatitis B virus by the CRISPR/Cas9 system via targeting the conserved regions of the viral genome. J. Gen. Virol. 2015, 96, 2252–2261. [Google Scholar] [CrossRef] [PubMed]
  75. Nishimasu, H.; Ran, F.A.; Hsu, P.D.; Konermann, S.; Shehata, S.I.; Dohmae, N.; Ishitani, R.; Zhang, F.; Nureki, O. Crystal Structure of Cas9 in Complex with Guide RNA and Target DNA. Cell 2014, 156, 935–949. [Google Scholar] [CrossRef] [Green Version]
  76. Feng, W.; Newbigging, A.M.; Tao, J.; Cao, Y.; Peng, H.; Le, C.; Wu, J.; Pang, B.; Li, J.; Tyrrell, D.L.; et al. CRISPR technology incorporating amplification strategies: Molecular assays for nucleic acids, proteins, and small molecules. Chem. Sci. 2021, 12, 4683–4698. [Google Scholar] [CrossRef]
  77. Abbott, T.R.; Dhamdhere, G.; Liu, Y.; Lin, X.; Goudy, L.; Zeng, L.; Chemparathy, A.; Chmura, S.; Heaton, N.S.; Debs, R.; et al. Development of CRISPR as an Antiviral Strategy to Combat SARS-CoV-2 and Influenza. Cell 2020, 181, 865–876.e12. [Google Scholar] [CrossRef]
  78. Woolhouse, M.E.J.; Brierley, L. Epidemiological characteristics of human-infective RNA viruses. Sci. Data 2018, 5, 180017. [Google Scholar] [CrossRef]
  79. Woolhouse, M.; Scott, F.; Hudson, Z.; Howey, R.; Chase-Topping, M. Human viruses: Discovery and emergence. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2012, 367, 2864–2871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Liu, L.; Li, X.; Ma, J.; Li, Z.; You, L.; Wang, J.; Wang, M.; Zhang, X.; Wang, Y. The Molecular Architecture for RNA-Guided RNA Cleavage by Cas13a. Cell 2017, 170, 714–726.e10. [Google Scholar] [CrossRef] [PubMed]
  81. Freije, C.A.; Myhrvold, C.; Boehm, C.K.; Lin, A.E.; Welch, N.L.; Carter, A.; Metsky, H.C.; Luo, C.Y.; Abudayyeh, O.O.; Gootenberg, J.S.; et al. Programmable Inhibition and Detection of RNA Viruses Using Cas13. Mol. Cell 2019, 76, 826–837.e11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ebina, H.; Misawa, N.; Kanemura, Y.; Koyanagi, Y. Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus. Sci. Rep. 2013, 3, 2510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hu, W.; Kaminski, R.; Yang, F.; Zhang, Y.; Cosentino, L.; Li, F.; Luo, B.; Alvarez-Carbonell, D.; Garcia-Mesa, Y.; Karn, J.; et al. RNA-directed gene editing specifically eradicates latent and prevents new HIV-1 infection. Proc. Natl. Acad. Sci. USA 2014, 111, 11461–11466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Yin, C.; Zhang, T.; Li, F.; Yang, F.; Putatunda, R.; Young, W.-B.; Khalili, K.; Hu, W.; Zhang, Y. Functional screening of guide RNAs targeting the regulatory and structural HIV-1 viral genome for a cure of AIDS. AIDS 2016, 30, 1163–1174. [Google Scholar] [CrossRef] [Green Version]
  85. Bayoumi, M.; Munir, M. Potential Use of CRISPR/Cas13 Machinery in Understanding Virus–Host Interaction. Front. Microbiol. 2021, 12, 743580. [Google Scholar] [CrossRef] [PubMed]
  86. Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.B.T.; Kellner, M.J.; Regev, A.; et al. RNA targeting with CRISPR–Cas13. Nature 2017, 550, 280–284. [Google Scholar] [CrossRef] [Green Version]
  87. Myhrvold, C.; Freije, C.A.; Gootenberg, J.S.; Abudayyeh, O.O.; Metsky, H.C.; Durbin, A.F.; Kellner, M.J.; Tan, A.L.; Paul, L.M.; Parham, L.A.; et al. Field-deployable viral diagnostics using CRISPR-Cas13. Science 2018, 360, 444–448. [Google Scholar] [CrossRef] [Green Version]
  88. Konermann, S.; Lotfy, P.; Brideau, N.J.; Oki, J.; Shokhirev, M.N.; Hsu, P.D. Transcriptome Engineering with RNA-Targeting Type VI-D CRISPR Effectors. Cell 2018, 173, 665–676.e14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Blanchard, E.L.; Vanover, D.; Bawage, S.S.; Tiwari, P.M.; Rotolo, L.; Beyersdorf, J.; Peck, H.E.; Bruno, N.C.; Hincapie, R.; Michel, F.; et al. Treatment of influenza and SARS-CoV-2 infections via mRNA-encoded Cas13a in rodents. Nat. Biotechnol. 2021, 39, 717–726. [Google Scholar] [CrossRef] [PubMed]
  90. Bawage, S.S.; Tiwari, P.M.; Santangelo, P.J. Synthetic mRNA expressed Cas13a mitigates RNA virus infections. bioRxiv 2018. [Google Scholar] [CrossRef] [Green Version]
  91. Ai, Y.; Liang, D.; Wilusz, J.E. CRISPR/Cas13 effectors have differing extents of off-target effects that limit their utility in eukaryotic cells. Nucleic Acids Res. 2022, 50, e65. [Google Scholar] [CrossRef]
  92. Hübner, A.; Petersen, B.; Keil, G.M.; Niemann, H.; Mettenleiter, T.C.; Fuchs, W. Efficient inhibition of African swine fever virus replication by CRISPR/Cas9 targeting of the viral p30 gene (CP204L). Sci. Rep. 2018, 8, 1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Roehm, P.C.; Shekarabi, M.; Wollebo, H.S.; Bellizzi, A.; He, L.; Salkind, J.; Khalili, K. Inhibition of HSV-1 Replication by Gene Editing Strategy. Sci. Rep. 2016, 6, 23146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Xu, X.; Fan, S.; Zhou, J.; Zhang, Y.; Che, Y.; Cai, H.; Wang, L.; Guo, L.; Liu, L.; Li, Q. The mutated tegument protein UL7 attenuates the virulence of herpes simplex virus 1 by reducing the modulation of α-4 gene transcription. Virol. J. 2016, 13, 152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. van Diemen, F.R.; Kruse, E.M.; Hooykaas, M.J.G.; Bruggeling, C.E.; Schürch, A.C.; van Ham, P.M.; Imhof, S.M.; Nijhuis, M.; Wiertz, E.J.H.J.; Lebbink, R.J. CRISPR/Cas9-Mediated Genome Editing of Herpesviruses Limits Productive and Latent Infections. PLoS Pathog. 2016, 12, e1005701. [Google Scholar] [CrossRef] [PubMed]
  96. Karpov, D.S.; Karpov, V.L.; Klimova, R.R.; Demidova, N.A.; Kushch, A.A. A Plasmid-Expressed CRISPR/Cas9 System Suppresses Replication of HSV Type I in a Vero Cell Culture. Mol. Biol. 2019, 53, 70–78. [Google Scholar] [CrossRef]
  97. Yin, D.; Ling, S.; Wang, D.; Dai, Y.; Jiang, H.; Zhou, X.; Paludan, S.R.; Hong, J.; Cai, Y. Targeting herpes simplex virus with CRISPR–Cas9 cures herpetic stromal keratitis in mice. Nat. Biotechnol. 2021, 39, 567–577. [Google Scholar] [CrossRef] [PubMed]
  98. Kaminski, R.; Bella, R.; Yin, C.; Otte, J.; Ferrante, P.; Gendelman, H.E.; Li, H.; Booze, R.; Gordon, J.; Hu, W.; et al. Excision of HIV-1 DNA by gene editing: A proof-of-concept in vivo study. Gene Ther. 2016, 23, 690–695. [Google Scholar] [CrossRef]
  99. Kaminski, R.; Chen, Y.; Fischer, T.; Tedaldi, E.; Napoli, A.; Zhang, Y.; Karn, J.; Hu, W.; Khalili, K. Elimination of HIV-1 Genomes from Human T-lymphoid Cells by CRISPR/Cas9 Gene Editing. Sci. Rep. 2016, 6, 22555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Wang, Q.; Liu, S.; Liu, Z.; Ke, Z.; Li, C.; Yu, X.; Chen, S.; Guo, D. Genome scale screening identification of SaCas9/gRNAs for targeting HIV-1 provirus and suppression of HIV-1 infection. Virus Res. 2018, 250, 21–30. [Google Scholar] [CrossRef] [PubMed]
  101. Yin, C.; Zhang, T.; Qu, X.; Zhang, Y.; Putatunda, R.; Xiao, X.; Li, F.; Xiao, W.; Zhao, H.; Dai, S.; et al. In Vivo Excision of HIV-1 Provirus by saCas9 and Multiplex Single-Guide RNAs in Animal Models. Mol. Ther. 2017, 25, 1168–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Herskovitz, J.; Hasan, M.; Patel, M.; Blomberg, W.R.; Cohen, J.D.; Machhi, J.; Shahjin, F.; Mosley, R.L.; McMillan, J.; Kevadiya, B.D.; et al. CRISPR-Cas9 Mediated Exonic Disruption for HIV-1 Elimination. EBioMedicine 2021, 73, 103678. [Google Scholar] [CrossRef] [PubMed]
  103. Yin, L.; Zhao, F.; Sun, H.; Wang, Z.; Huang, Y.; Zhu, W.; Xu, F.; Mei, S.; Liu, X.; Zhang, D.; et al. CRISPR-Cas13a Inhibits HIV-1 Infection. Mol. Ther. Nucleic Acids 2020, 21, 147–155. [Google Scholar] [CrossRef] [PubMed]
  104. Dong, C.; Qu, L.; Wang, H.; Wei, L.; Dong, Y.; Xiong, S. Targeting hepatitis B virus cccDNA by CRISPR/Cas9 nuclease efficiently inhibits viral replication. Antivir. Res. 2015, 118, 110–117. [Google Scholar] [CrossRef] [PubMed]
  105. Kostyushev, D.; Brezgin, S.; Kostyusheva, A.; Zarifyan, D.; Goptar, I.; Chulanov, V. Orthologous CRISPR/Cas9 systems for specific and efficient degradation of covalently closed circular DNA of hepatitis B virus. Cell. Mol. Life Sci. 2019, 76, 1779–1794. [Google Scholar] [CrossRef] [PubMed]
  106. Yan, K.; Feng, J.; Liu, X.; Wang, H.; Li, Q.; Li, J.; Xu, T.; Sajid, M.; Ullah, H.; Zhou, L.; et al. Inhibition of Hepatitis B Virus by AAV8-Derived CRISPR/SaCas9 Expressed From Liver-Specific Promoters. Front. Microbiol. 2021, 12, 665184. [Google Scholar] [CrossRef] [PubMed]
  107. Tang, Y.-D.; Liu, J.-T.; Wang, T.-Y.; Sun, M.-X.; Tian, Z.-J.; Cai, X.-H. CRISPR/Cas9-mediated multiple single guide RNAs potently abrogate pseudorabies virus replication. Arch. Virol. 2017, 162, 3881–3886. [Google Scholar] [CrossRef]
  108. Jubair, L.; Fallaha, S.; McMillan, N.A.J. Systemic Delivery of CRISPR/Cas9 Targeting HPV Oncogenes Is Effective at Eliminating Established Tumors. Mol. Ther. 2019, 27, 2091–2099. [Google Scholar] [CrossRef] [PubMed]
  109. Zhen, S.; Hua, L.; Takahashi, Y.; Narita, S.; Liu, Y.-H.; Li, Y. In vitro and in vivo growth suppression of human papillomavirus 16-positive cervical cancer cells by CRISPR/Cas9. Biochem. Biophys. Res. Commun. 2014, 450, 1422–1426. [Google Scholar] [CrossRef] [PubMed]
  110. Gao, X.; Jin, Z.; Tan, X.; Zhang, C.; Zou, C.; Zhang, W.; Ding, J.; Das, B.C.; Severinov, K.; Hitzeroth, I.I.; et al. Hyperbranched poly(β-amino ester) based polyplex nanopaticles for delivery of CRISPR/Cas9 system and treatment of HPV infection associated cervical cancer. J. Control. Release 2020, 321, 654–668. [Google Scholar] [CrossRef] [PubMed]
  111. Yoshiba, T.; Saga, Y.; Urabe, M.; Uchibor, R.; Matsubara, S.; Fujiwara, H.; Mizukami, H. CRISPR/Cas9-mediated cervical cancer treatment targeting human papillomavirus E6. Oncol Lett. 2019, 17, 2197–2206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kennedy, E.M.; Kornepati, A.V.R.; Goldstein, M.; Bogerd, H.P.; Poling, B.C.; Whisnant, A.W.; Kastan, M.B.; Cullen, B.R. Inactivation of the human papillomavirus E6 or E7 gene in cervical carcinoma cells by using a bacterial CRISPR/Cas RNA-guided endonuclease. J. Virol. 2014, 88, 11965–11972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Wang, L.; Zhou, J.; Wang, Q.; Wang, Y.; Kang, C. Rapid design and development of CRISPR-Cas13a targeting SARS-CoV-2 spike protein. Theranostics 2021, 11, 649–664. [Google Scholar] [CrossRef]
  114. Nguyen, T.M.; Zhang, Y.; Pandolfi, P.P. Virus against virus: A potential treatment for 2019-nCov (SARS-CoV-2) and other RNA viruses. Cell Res. 2020, 30, 189–190. [Google Scholar] [CrossRef] [Green Version]
  115. Cui, J.; Techakriengkrai, N.; Nedumpun, T.; Suradhat, S. Abrogation of PRRSV infectivity by CRISPR-Cas13b-mediated viral RNA cleavage in mammalian cells. Sci. Rep. 2020, 10, 9617. [Google Scholar] [CrossRef]
  116. Kwon, M.H.; Lee, M.S.; Kim, K.H.; Park, S.; Shin, H.J.; Jang, Y.J.; Kim, H.I. Production and characterization of an anti-idiotypic single chain Fv that recognizes an anti-DNA antibody. Immunol. Investig. 2002, 31, 205–218. [Google Scholar] [CrossRef]
  117. Kim, Y.R.; Kim, J.S.; Lee, S.H.; Lee, W.R.; Sohn, J.N.; Chung, Y.C.; Shim, H.K.; Lee, S.C.; Kwon, M.H.; Kim, Y.S. Heavy and light chain variable single domains of an anti-DNA binding antibody hydrolyze both double- and single-stranded DNAs without sequence specificity. J. Biol. Chem. 2006, 281, 15287–15295. [Google Scholar] [CrossRef] [Green Version]
  118. Lee, G.; Cho, S.; Hoang, P.M.; Kim, D.; Lee, Y.; Kil, E.J.; Byun, S.J.; Lee, T.K.; Kim, D.H.; Kim, S.; et al. Therapeutic Strategy for the Prevention of Pseudorabies Virus Infection in C57BL/6 Mice by 3D8 scFv with Intrinsic Nuclease Activity. Mol. Cells 2015, 38, 773–780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Lee, G.; Yu, J.; Cho, S.; Byun, S.J.; Kim, D.H.; Lee, T.K.; Kwon, M.H.; Lee, S. A nucleic-acid hydrolyzing single chain antibody confers resistance to DNA virus infection in hela cells and C57BL/6 mice. PLoS Pathog. 2014, 10, e1004208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Lee, Y.; Hoang, P.T.; Kim, D.; Ayun, R.Q.; Luong, Q.X.T.; Na, K.; Kim, T.; Oh, Y.; Kim, W.-K.; Lee, S. A Therapeutically Active Minibody Exhibits an Antiviral Activity in Oseltamivir-Resistant Influenza-Infected Mice via Direct Hydrolysis of Viral RNAs. Viruses 2022, 14, 1105. [Google Scholar] [CrossRef]
  121. Hoang, P.M.; Cho, S.; Kim, K.E.; Byun, S.J.; Lee, T.K.; Lee, S. Development of Lactobacillus paracasei harboring nucleic acid-hydrolyzing 3D8 scFv as a preventive probiotic against murine norovirus infection. Appl. Microbiol. Biotechnol. 2015, 99, 2793–2803. [Google Scholar] [CrossRef] [PubMed]
  122. Jung, Y.; Rhee, Y.; Auh, C.K.; Shim, H.; Choi, J.J.; Kwon, S.T.; Yang, J.S.; Kim, D.; Kwon, M.H.; Kim, Y.S.; et al. Production of recombinant single chain antibodies (scFv) in vegetatively reproductive Kalanchoe pinnata by in planta transformation. Plant Cell Rep. 2009, 28, 1593–1602. [Google Scholar] [CrossRef] [PubMed]
  123. Lee, J.; Kim, M.; Seo, Y.; Lee, Y.; Park, H.; Byun, S.J.; Kwon, M.H. The catalytic activity of a recombinant single chain variable fragment nucleic acid-hydrolysing antibody varies with fusion tag and expression host. Arch. Biochem. Biophys. 2017, 633, 110–117. [Google Scholar] [CrossRef]
  124. Park, H.; Kim, M.; Kim, H.J.; Lee, Y.; Seo, Y.; Pham, C.D.; Lee, J.; Byun, S.J.; Kwon, M.H. Heparan sulfate proteoglycans (HSPGs) and chondroitin sulfate proteoglycans (CSPGs) function as endocytic receptors for an internalizing anti-nucleic acid antibody. Sci. Rep. 2017, 7, 14373. [Google Scholar] [CrossRef] [PubMed]
  125. Jang, J.Y.; Jeong, J.G.; Jun, H.R.; Lee, S.C.; Kim, J.S.; Kim, Y.S.; Kwon, M.H. A nucleic acid-hydrolyzing antibody penetrates into cells via caveolae-mediated endocytosis, localizes in the cytosol and exhibits cytotoxicity. Cell Mol. Life Sci. 2009, 66, 1985–1997. [Google Scholar] [CrossRef]
  126. Cho, S.; Youn, H.N.; Hoang, P.M.; Cho, S.; Kim, K.E.; Kil, E.J.; Lee, G.; Cho, M.J.; Hong, J.; Byun, S.J.; et al. Preventive Activity against Influenza (H1N1) Virus by Intranasally Delivered RNA-Hydrolyzing Antibody in Respiratory Epithelial Cells of Mice. Viruses 2015, 7, 5133–5144. [Google Scholar] [CrossRef]
  127. Jun, H.R.; Pham, C.D.; Lim, S.I.; Lee, S.C.; Kim, Y.S.; Park, S.; Kwon, M.H. An RNA-hydrolyzing recombinant antibody exhibits an antiviral activity against classical swine fever virus. Biochem. Biophys. Res. Commun. 2010, 395, 484–489. [Google Scholar] [CrossRef] [Green Version]
  128. Park, J.H.; Lee, J.W.; Choi, H.; Jung, S.K.; Kim, J.S.; Kim, K.W.; Oh, K.B.; Yang, H.; Byun, S.J. Survival of Escherichia coli harboring nucleic acid-hydrolyzing 3D8 scFv during RNA virus infection. Regul. Toxicol. Pharmacol. 2018, 94, 286–292. [Google Scholar] [CrossRef]
  129. Sureshkumar, S.; Jung, S.K.; Kim, D.; Oh, K.B.; Yang, H.; Lee, H.C.; Jo, Y.J.; Lee, H.S.; Lee, S.; Byun, S.J. Administration of L. salivarius expressing 3D8 scFv as a feed additive improved the growth performance, immune homeostasis, and gut microbiota of chickens. Anim. Sci. J. 2020, 91, e13399. [Google Scholar] [CrossRef] [PubMed]
  130. Sureshkumar, S.; Jung, S.K.; Kim, D.; Oh, K.B.; Yang, H.; Lee, H.C.; Jin, J.Y.; Sun, L.H.; Lee, S.; Byun, S.J. Oral administration of Lactobacillus reuteri expressing a 3D8 single-chain variable fragment (scFv) enhances chicken growth and conserves immune homeostasis. 3 Biotech. 2019, 9, 282. [Google Scholar] [CrossRef]
  131. Cho, S.; Kim, D.; Lee, Y.; Kil, E.J.; Cho, M.J.; Byun, S.J.; Cho, W.K.; Lee, S. Probiotic Lactobacillus Paracasei Expressing a Nucleic Acid-Hydrolyzing Minibody (3D8 Scfv) Enhances Probiotic Activities in Mice Intestine as Revealed by Metagenomic Analyses. Genes 2018, 9, 276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Kim, D.S.; Lee, S.H.; Kim, J.S.; Lee, S.C.; Kwon, M.H.; Kim, Y.S. Generation of humanized anti-DNA hydrolyzing catalytic antibodies by complementarity determining region grafting. Biochem. Biophys. Res. Commun. 2009, 379, 314–318. [Google Scholar] [CrossRef] [PubMed]
  133. Roh, J.; Byun, S.J.; Seo, Y.; Kim, M.; Lee, J.H.; Kim, S.; Lee, Y.; Lee, K.W.; Kim, J.K.; Kwon, M.H. Generation of a chickenized catalytic anti-nucleic acid antibody by complementarity-determining region grafting. Mol. Immunol. 2015, 63, 513–520. [Google Scholar] [CrossRef] [PubMed]
  134. Son, S.; Lee, W.R.; Joung, Y.K.; Kwon, M.H.; Kim, Y.S.; Park, K.D. Optimized stability retention of a monoclonal antibody in the PLGA nanoparticles. Int. J. Pharm. 2009, 368, 178–185. [Google Scholar] [CrossRef] [PubMed]
  135. Lee, G.; Shim, H.-K.; Kwon, M.-H.; Son, S.-H.; Kim, K.-Y.; Park, E.-Y.; Yang, J.-K.; Lee, T.-K.; Auh, C.-K.; Kim, D.; et al. RNA virus accumulation is inhibited by ribonuclease activity of 3D8 scFv in transgenic Nicotiana tabacum. Plant Cell Tissue Organ. Cult. (PCTOC) 2013, 115, 189–197. [Google Scholar] [CrossRef]
  136. Tran, D.T.; Cho, S.; Hoang, P.M.; Kim, J.; Kil, E.-J.; Lee, T.-K.; Rhee, Y.; Lee, S. A Codon-Optimized Nucleic Acid Hydrolyzing Single-Chain Antibody Confers Resistance to Chrysanthemums Against Chrysanthemum Stunt Viroid Infection. Plant Mol. Biol. Rep. 2016, 34, 221–232. [Google Scholar] [CrossRef]
  137. June Byun, S.; Yuk, S.-s.; Jang, Y.-J.; Choi, H.; Jeon, M.-H.; Erdene-Ochir, T.O.; Kwon, J.-H.; Noh, J.-Y.; Sun Kim, J.; Gyu Yoo, J.; et al. Transgenic Chickens Expressing the 3D8 Single Chain Variable Fragment Protein Suppress Avian Influenza Transmission. Sci. Rep. 2017, 7, 5938. [Google Scholar] [CrossRef]
  138. Lee, G.; Choi, H.; Sureshkumar, S.; Jung, S.K.; Kim, J.S.; Oh, K.B.; Kim, K.W.; Yang, H.; Kim, D.H.; Byun, S.J. The 3D8 single chain variable fragment protein suppress infectious bronchitis virus transmission in the transgenic chickens. Res. Vet. Sci. 2019, 123, 293–297. [Google Scholar] [CrossRef]
  139. Byun, S.J.; Choi, H.; Sureshkumar, S.; Yuk, S.-S.; Kwon, J.-H.; Noh, J.-Y.; Jung, S.K.; Kim, J.S.; Oh, K.B.; Yang, H.; et al. The 3D8 single chain variable fragment protein suppresses Newcastle disease virus transmission in transgenic chickens. BMC Vet. Res. 2020, 16, 273. [Google Scholar] [CrossRef] [PubMed]
  140. Lee, G.; Budhathoki, S.; Lee, G.Y.; Oh, K.J.; Ham, Y.K.; Kim, Y.J.; Lim, Y.R.; Hoang, P.T.; Lee, Y.; Lim, S.W.; et al. Broad-Spectrum Antiviral Activity of 3D8, a Nucleic Acid-Hydrolyzing Single-Chain Variable Fragment (scFv), Targeting SARS-CoV-2 and Multiple Coronaviruses In Vitro. Viruses 2021, 13, 650. [Google Scholar] [CrossRef] [PubMed]
  141. Lee, G.; Shim, H.-K.; Kwon, M.-H.; Son, S.-H.; Kim, K.-Y.; Park, E.-Y.; Lee, T.-K.; Lee, W.-R.; Auh, C.-K.; Kim, D.; et al. A nucleic acid hydrolyzing recombinant antibody confers resistance to curtovirus infection in tobacco. Plant Cell Tissue Organ. Cult. (PCTOC) 2013, 115, 179–187. [Google Scholar] [CrossRef]
  142. Xu, C.; Zhou, Y.; Xiao, Q.; He, B.; Geng, G.; Wang, Z.; Cao, B.; Dong, X.; Bai, W.; Wang, Y.; et al. Programmable RNA editing with compact CRISPR–Cas13 systems from uncultivated microbes. Nat. Methods 2021, 18, 499–506. [Google Scholar] [CrossRef]
Figure 1. Proposed mechanism of viral genome degradation-specific antiviral candidates. (A) Typical viral life cycle of enveloped viruses; virus infection to host cells includes several main steps, 1. Binding; 2. Fusion; 3. Release of viral genome in the cytoplasm; 4a. Replication in the cytoplasmic or 4b in the nucleus (viral genome needs to be transported to the nucleus), 5. Transcription and presence of mRNA in the cytoplasm for translation; 6. Viral genome replication; 7a, 7b. Assembly and release of new virions. (B) 3D8 scFv with cell penetration ability via caveolar endocytosis and localization in the cytoplasm, intracellular localization of other candidates (CRISPR/Cas, ISG20, RNases, and RNAi) are proposed with a delivery system and expressed as protein inside the cells. (C) The presence of viral genomes cleavage candidates (3D8 scFv, CRISPR/Cas, ISG20, RNases, RNAi) inhibits viral replication. Upon infection, viral genomes distributed in the cytoplasm were digested by RNase, ISG20, CRISPR/Cas, 3D8 scFv, and RNAi. 3D8 scFv, RNAi, and CRISPR/Cas were supposed to degrade viral mRNA in the cytoplasm for translation. Finally, the replicated viral genomes can be catalyzed using 3D8 scFv, RNAi, and CRISPR/Cas.
Figure 1. Proposed mechanism of viral genome degradation-specific antiviral candidates. (A) Typical viral life cycle of enveloped viruses; virus infection to host cells includes several main steps, 1. Binding; 2. Fusion; 3. Release of viral genome in the cytoplasm; 4a. Replication in the cytoplasmic or 4b in the nucleus (viral genome needs to be transported to the nucleus), 5. Transcription and presence of mRNA in the cytoplasm for translation; 6. Viral genome replication; 7a, 7b. Assembly and release of new virions. (B) 3D8 scFv with cell penetration ability via caveolar endocytosis and localization in the cytoplasm, intracellular localization of other candidates (CRISPR/Cas, ISG20, RNases, and RNAi) are proposed with a delivery system and expressed as protein inside the cells. (C) The presence of viral genomes cleavage candidates (3D8 scFv, CRISPR/Cas, ISG20, RNases, RNAi) inhibits viral replication. Upon infection, viral genomes distributed in the cytoplasm were digested by RNase, ISG20, CRISPR/Cas, 3D8 scFv, and RNAi. 3D8 scFv, RNAi, and CRISPR/Cas were supposed to degrade viral mRNA in the cytoplasm for translation. Finally, the replicated viral genomes can be catalyzed using 3D8 scFv, RNAi, and CRISPR/Cas.
Microorganisms 10 01552 g001
Table 1. The global epidemic and pandemic of viral diseases throughout history.
Table 1. The global epidemic and pandemic of viral diseases throughout history.
No.Year 1DiseaseVirusDeath 2
11520SmallpoxVariola virus500 million
2~1800Yellow feverYellow fever virus>210,000
31918Spanish fluInfluenza A virus (H1N1)50 million
41957Asian fluInfluenza A virus (H2N2)2 million
51968Hong Kong fluInfluenza A virus (H3N2)1 million
61976EbolaEbola virus~15,300
71981HIV/AIDSHIV~37 million
81990Dengue feverDengue virus>680,000
92002SARSSARS-CoV774
102009Swine fluInfluenza A virus (H1N1)284,000
112012MERSMERS-CoV891
122014ChikungunyaChikungunya virusrare
132015ZikaZika virus~1000
142020COVID-19SARS-CoV-2~6.2 million
1 Refer to the year of the first outbreak case, 2 refer to the estimated number of deaths until recently.
Table 4. 3D8 scFv hydrolyzes viral genomes exhibiting antiviral activity against broad-spectrum viruses.
Table 4. 3D8 scFv hydrolyzes viral genomes exhibiting antiviral activity against broad-spectrum viruses.
DerivationExperimentVirusGenomeReplicationGenusFamilyRef.
Protein expressed in E. coliPK-15 cellsCSFV(+)ssRNACytoplasmPestivirusFlaviviridae[127]
Transgenic cell line
Protein expressed in E. coliHela cellsVSV(−)ssRNACytoplasmVesiculovirusRhabdoviridae[134]
Transgenic plantsN. tabacumPMMoV(+)ssRNACytoplasmTobamovirusVirgaviridae[135]
TMGMV
ToMV
TMV
CMVCucumovirusBromoviridae[135]
Transgenic plantsChrysanthemumsCSVd(−)ssRNANucleusPospiviroidPospiviroidae[136]
Protein expressed in L. paracaseRAW264.7 cellsMNV1(+)ssRNACytoplasmNorovirusCalciviridae[121]
Transgenic bacteria L. paracaseMice
Protein expressed in E. coliMDCK cellH1N1/NWS33(−)ssRNANucleusInfluenzavirus AOrthomyxoviridae[120]
H9N2
H1N1/PR8
H3N2
MDCK cell/MiceH1N1/09pdm
Transgenic animalChickensH9N2(−)ssRNANucleusInfluenzavirus AOrthomyxoviridae[137]
Transgenic animalInfectious bronchitis virus(+)ssRNACytoplasmGammacoronavirusCoronaviridae[138]
Transgenic animalNewcastle disease(−)ssRNACytoplasmAvulavirusParamyxoviridae[139]
Protein expressed in E. coliVero E6SARS-CoV-2(+)ssRNACytoplasmBetacoronavirusCoronaviridae[140]
hCo-OC43BetacoronavirusCoronaviridae
PEDVAlphacoronavirusCoronaviridae
Transgenic cell lineHelaHSV1dsDNANucleusSimplexvirusHerpesviridae[119]
Transgenic cell linePRVdsDNANucleusVaricellovirusHerpesviridae[119]
Transgenic animalMicedsDNANucleusVaricellovirusHerpesviridae[118]
Transgenic plantsN. tabacumBCTVssDNANucleusCurtovirusGeminiviridae[141]
Transgenic plantsN. tabacumBSCTV
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hoang, P.T.; Luong, Q.X.T.; Ayun, R.Q.; Lee, Y.; Vo, T.T.B.; Kim, T.; Lee, S. A Novel Approach of Antiviral Drugs Targeting Viral Genomes. Microorganisms 2022, 10, 1552. https://doi.org/10.3390/microorganisms10081552

AMA Style

Hoang PT, Luong QXT, Ayun RQ, Lee Y, Vo TTB, Kim T, Lee S. A Novel Approach of Antiviral Drugs Targeting Viral Genomes. Microorganisms. 2022; 10(8):1552. https://doi.org/10.3390/microorganisms10081552

Chicago/Turabian Style

Hoang, Phuong Thi, Quynh Xuan Thi Luong, Ramadhani Qurrota Ayun, Yongjun Lee, Thuy Thi Bich Vo, Taehyun Kim, and Sukchan Lee. 2022. "A Novel Approach of Antiviral Drugs Targeting Viral Genomes" Microorganisms 10, no. 8: 1552. https://doi.org/10.3390/microorganisms10081552

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop